Nonfunctioning Pancreatic Neuroendocrine Tumors


ENETS TNM

UICC/AJCC/WHO 2010 TNM

T classification

Limited to the pancreas, <2 cm

Limited to the pancreas, ≤2 cm in greatest dimension

Limited to the pancreas, 2–4 cm

Limited to the pancreas, >2 cm in greatest dimension

Limited to the pancreas, >4 cm or invading duodenum or bile duct

Beyond the pancreas but without involvement of the superior mesenteric artery

Tumor invading adjacent organs (stomach, spleen, colon, adrenal gland) or the wall of large vessels (celiac axis or the superior mesenteric artery)

Involvement of celiac axis or the superior mesenteric artery (unresectable tumor)

N classification

No regional lymph node metastasis

No regional lymph node metastasis

Regional lymph node metastasis

Regional lymph node metastasis

M classification

No distant metastasis

No distant metastasis

Distant metastasis

Distant metastasis

Stage classification

I: T1, N0, M0

IA: T1, N0, M0

IB: T2, N0, M0

IIa: T2, N0, M0

IIA: T3, N0, M0

IIb: T3, N0, M0

IIB: T1–T3, N1, M0

IIIa: T4, N0, M0

III: T4, N0–N1, M0

IIIb: T1–T4, N1, M0

IV: T1–T4, N0–N1, M1

IV: T1–T4, N0–N1, M1


Data from Rindi et al. [92, 93], Bosman et al. [9], Edge et al. [25]

Abbreviations: NA not applicable





18.4 Prognostic and Predictive Factors


Over the past few years, a few studies have identified certain clinical parameters that may contribute to the formation of an accurate prognostic assessment, which is of paramount importance for therapeutic decision-making. The most prognostically important pathological factors are the presence of angioinvasion as well as the proliferation index [59, 89]. With respect to the latter, the Ki-67 labeling index emerges as an independent predictor of survival using a cutoff value of 5 % in both functioning and NF-PNETs [96]. Together with G3 histology and the proliferative index Ki-67, with an increasing risk of progression of 2 % for each increasing Ki-67 unit, are the major factors to predict tumor progression [86]. One Italian study evaluating prognostic factors in NF-PNETs including 180 patients supports that the evaluation of nodal metastasis and weight loss should be included along with liver metastasis, Ki-67 index, and grade of differentiation [7]. One of the largest studies in the literature evaluating 2,158 NF-PNETs showed that systemic metastases, local/vascular/lymphatic invasion, and grade are more powerful indicators of outcome with tumor grade influencing survival more than distant metastases [38].

It is well established that radical surgery is the only chance for cure in NF-PNETs, with a 5-year survival rate of 93 % [7]. Nevertheless, the surgical choice is the direct result of the disease stage at diagnosis, a well-established prognostic factor in all tumors.

With respect to predictive factors and response to chemotherapy, this was found to be related to Ki-67 LI >10 %, the performance status, and age <60 years [108]. Additionally, a recent study showed that apart from a high Ki-67 LI >5 %, the lymph node ratio in resected PNETs is a powerful predictor of recurrence and supports its investigation in relation to adjuvant treatment in the context of clinical trials [8].


18.5 Therapies



18.5.1 Surgical Therapy


Surgical resection remains the cornerstone therapy and the only approach that may cure patients with G1 and G2 PNETs. With respect to G3 tumors, surgery is rarely curative but offers a significant long-term palliation [34].

Over the last decade, novel and less invasive pancreatic surgical approaches have been developed and depending on clinical scenarios are applied in patients with PNETs [41]. These involve mainly partial pancreatectomies or tumor enucleation that may also be performed laparoscopically [34]. Generally, typical and atypical pancreatic resections are intended for different sites and sizes of PNETs, and the decision should be ideally based on previous histological confirmation by endoscopic ultrasound. Tumor enucleation has been proposed in well-demarcated, less than 2 cm, and benign or uncertain-behavior lesions [32]. Centrally located lesions of the pancreatic head require pancreaticoduodenectomy, while lesions of the body and tail may be handled with a left pancreatectomy with or without spleen preservation. Middle pancreatectomy is usually performed for small tumors of the pancreatic body, while enucleation is considered when the main pancreatic duct can be preserved.

The role of surgical resection of the primary PNET in the presence of inoperable metastases is controversial. In 2,158 NF-PNET patients from the SEER database, it was shown that removal of the primary tumor significantly prolonged survival in the entire cohort (median 1.2 vs. 8.4 years; p < 0.001) even among those with metastatic disease (median 1.0 vs. 4.8 years; p < 0.001) [38]. On the other hand, a systematic review of small studies specifically investigating the role of surgery in this scenario was inconclusive supporting the need of a prospective randomized trial [11]. Furthermore, no survival difference was seen between enucleation and more aggressive resection of the primary tumor (median 10.2 vs. 9.2 years, p = 0.456) [38].

Commonly, typical pancreatectomies are associated with a high rate of perioperative complications as well as exocrine and endocrine insufficiency [98]. Atypical resections, on the other hand, are associated with a decreased long-term endocrine/exocrine impairment but with a high incidence of pancreatic fistulas, a disturbing but ultimately a manageable complication [2, 31].

Whichever the type of resection, for a more than 2 cm suspicious lesion, it is imperative to perform a wide excision with clear margins and to remove draining lymph nodes for proper TNM assessment particularly in cases with G2 and G3 PNETs. Controversial is the management of tumors 1–2 cm and those found incidentally as several studies have shown that these may be malignant and have lymph nodal metastases [35, 42, 88]. It is recommended depending on each case either to closely monitor or to remove with draining lymph nodes.


18.5.1.1 Surgical Therapy of Patients with NF-PNETs in the Context of MEN1


MEN1 syndrome represents a difficult challenge for surgeons mainly because it is associated with heterochronous and multifocal NF-PNETs. Prophylactic pancreatectomies could remove all premalignant lesions prior to malignant conversion but are not an accepted approach. Therefore, the careful monitoring of these patients for the early diagnosis and surgical excision of any arising tumors with malignant potential is of utmost importance [55]. Based on previous evidence, the European Neuroendocrine Tumor Society (ENETS) recommends operating MEN1-related NF-PNETs with a diameter of more than 2 cm, with a yearly size increment in diameter of more than 0.5 cm, or with metastases [30, 105]. The management of NF-PNETs of less than 2 cm is debated as they may also have an aggressive biology, but they usually are microadenomas and have a more indolent behavior, and their management is close monitoring.

Overall, the refinement and personalization of surgical procedures in NF-PNETs have resulted in improved long-term survival in patients with locally advanced and metastatic disease, as well as shorter hospital stays and comparable long-term outcomes in patients with limited disease treated minimally invasively. There are still controversies related to issues of surgical treatment of PNETs, such as to what extent enucleation, lymph node sampling and vascular reconstruction are beneficial for the oncologic outcome particularly in MEN1 patients and in tumors 1–2 cm in diameter. It is expected that the recently developed endoscopic ultrasound-guided biopsies will shed light to all these gray zones.


18.5.2 Medical Therapies


Recent advances in therapeutics have introduced novel exciting agents in the treatment of PNETs that are expected to improve the survival of patients. Some interesting combinations including triplets of chemotherapy, radiopeptides, or targeted agents have recently been exploited in this rapidly evolving field (Table 18.2).


Table 18.2
List of mainstream clinical studies in pancreatic neuroendocrine tumors

























































































































Drugs

N pts

Ki-67

SSTR+

PR %

PFSa/TTPb months

Study type

Author

STZ/ADM/5-FU

84

NR

NR

34

9a

Retrospective

Kouvaraki et al. [54]

TMZ

36

NR

NR

14

7b

Retrospective

Ekeblad et al. [26]

STZ/CDDP/5-FU

49

25

39

38

9b

Retrospective

Turner et al. [106]

TMZ/Xeloda

30

NR

NR

70

18a

Retrospective

Strosberg et al. [101]

TMZ/BEV

15

<20 %

NR

33

14.3a

Phase II

Chan et al. [15]

PRRT (Lu)

91

NR

91

43

40a

Retrospective

Kwekkeboom et al. [57]

PRRT (Lu)

52

NR

52

29

29a

Phase II

Sansovini et al. [95]

Sunitinib

86

36

NR

7

11.4a

Phase III

Raymond et al. [91]

Placebo

85

36
 
0

5.5a
   

Everolimus

207

NR

NR

5

11a

Phase III

Yao et al. [117]

Placebo

203
     
4.6a
   


STZ streptozotocin, ADM Adriamycin, TMZ temozolomide, XELOX Xeloda/oxaliplatin, CDDP cisplatin, 5-FU 5-fluorouracil, BEV bevacizumab, PRRT peptide receptor radionuclide therapy, Lu lutetium, SSTR somatostatin receptors, PR partial response, TTP time to progression, PFS progression-free survival, NR not reported

aProgressive free survival (PFS)

bTime to disease progression (TPP)


18.5.2.1 Somatostatin Analogs


The clinical application of long-acting somatostatin analogs (SSAs) has changed dramatically the quality of life and prognosis of patients with NETs [79]. Their benefit was initially thought to be related to their antisecretory effect as they were shown to relieve patients from troubling clinical syndromes related to the peptide secretion. Altogether, SSAs have been shown to be well tolerated and safe in all available forms as patients may experience only mild and infrequent side effects such as abdominal pain, steatorrhea, and cholelithiasis [81]. This last effect is due to the inhibitory effects of octreotide on gallbladder contractility, and although it requires follow-up attention, is rarely of clinical significance.

From the advent of the first SSA, an improved survival has been shown in NET patients, and although a clear gain could be seen by the fact that patients’ quality of life was improved together with their symptoms, it was not clear whether there was also an antiproliferative effect. Several groups have reported a possible antiproliferative action from SSA, by induction of apoptosis [1, 23, 44], although significant tumor shrinkage has been reported to occur in <5 % of cases [79]. Initial in vitro studies suggested a number of direct and indirect mechanisms of such an effect. Direct mechanisms involve the activation of somatostatin receptors (SSRs) leading to modulation of intracellular signaling transduction such as the activation of MAP kinase pathway [118]. Indirect antiproliferative effects are exerted through activation of SSRs on endothelial cells and monocytes leading to blocking of tumor angiogenesis [20, 49]. A proof of direct antiproliferative effect came from the PROMID trial, a phase III study that randomized patients to receive octreotide long-acting repeatable (LAR) or placebo [94]. This placebo-controlled, double-blind study involved patients with well-differentiated metastatic NETs originating in the distal intestine and proximal colon and showed a clear benefit in all patients receiving octreotide including those with NF tumors. Nevertheless, in this trial, disease stabilization was the main clinical response observed as hardly any tumor shrinkage was achieved and there were no patients with NF-PNETs included in this study. Furthermore, no clear prospective evidence supporting the antitumoral effect of SSAs in NF-PNETs exists. However, based on these encouraging results, octreotide LAR was recommended [84] and approved in several countries in progressing well-differentiated, metastatic nonfunctioning NETs, including PNETs. Supporting are the results of a randomized phase III double-blind trial testing lanreotide versus placebo in NF well-differentiated enteropancreatic NETs (the CLARINET study; clinical trial link: http://​clinicaltrials.​gov/​show/​NCT00353496). In a previous study including 21 well-differentiated advanced NF-PNETs, treatment with octreotide LAR was associated with disease stabilization and good quality of life in 38 % of patients [10]. A Ki-67 > or =5 % and/or weight loss was associated with more aggressive disease and would justify more aggressive therapy [10].


18.5.2.2 Interferon


Early studies had shown that interferon-alpha (IFN-α) has antiproliferative, through Jak1/Tyk2 kinase and STAT1/STAT2 activation, antisecretory, antiangiogenic, and antiapoptotic effects [22, 76, 109]. A possible positive immunomodulatory effect on T-cell and natural killer activity was also suggested [39, 77], but it has not been confirmed since by later studies. Interferon-α may be used as monotherapy or in combination with SSAs, as antisecretory and antiproliferative treatment, with variable response rates (RRs). There has been biochemical response in 40–60 % of patients, symptomatic improvement in 40–70 % of patients, and significant tumor shrinkage in a median of 10–15 % of patients [37, 78, 82]. Side effects of IFN-α are significant and include flu-like syndrome, chronic fatigue, depression, weight loss, polyneuropathy, myositis, thrombocytopenia, anemia, leukopenia, and autoantibodies in 15 % of patients (autoimmune hepatitis/thyroiditis, psoriasis, systemic lupus erythematosus syndrome with antinuclear factors, and parietal cell antibodies with pernicious anemia) [78, 84].

One of the first reports to compare results from different studies on octreotide and on IFN-α monotherapy in NETs suggested that better biochemical responses and disease stabilization were obtained with octreotide and not with IFN-α [17]. An initial single-arm study suggested that for patients with metastasized endocrine gastroenteropancreatic tumors, the combination of octreotide and IFN-α is superior to either as monotherapy [75]. Another early study showed that the addition of IFN-α to octreotide has antiproliferative efficacy in a subgroup of patients with advanced metastatic disease unresponsive to octreotide monotherapy [37]. Similarly, in another study, patients having progressed on either octreotide or IFN-α benefited from the subsequent combination of both drugs [36]. There are very few studies comparing SSA monotherapy to IFN/SSA combinations, and the results are conflicting. There are no studies employing IFN monotherapy or in combination with SSA in NF-PNETs. A prospective randomized study showed a significantly reduced risk of tumor progression during follow-up in patients with midgut carcinoid tumors metastatic to the liver receiving octreotide and IFN-α as compared to single-agent octreotide [50]. A randomized phase III study comprised three arms to compare lanreotide, IFN and lanreotide plus IFN [28]. All arms had equivalent results with respect to symptom control. No arm showed statistically significant survival improvement, but there was a trend of better survival in the combination arm. In the same study, the most toxic arm was the combination one. Another recent randomized trial comparing octreotide to octreotide/IFN-α showed no survival advantage between the two arms [4]. It is hypothesized that the difference in the results lies on the heterogeneity of the disease and further larger trials are required. At present, we do not have enough statistical evidence for an upfront use of the combination of IFN-α and SSA in patients with NETs, but we have some clinical evidence coming from non-randomized studies and the sub-analysis of randomized trials that would justify the sequential use of the two drugs or the combination after progression to single-agent therapy [33].

Head to head comparisons of octreotide/IFN-α to other biological combinations have not been performed yet. A randomized phase III trial is currently recruiting patients in order to compare the efficacy of octreotide/IFN-α2b and octreotide/bevacizumab in metastatic or locally advanced, high-risk NETs (clinical trial link: http://​clinicaltrials.​gov/​show/​NCT00569127). This trial was based on a previous phase II study showing better PFS and objective responses of octreotide/bevacizumab compared to octreotide/PEG-IFN [116].


18.5.2.3 Targeted Therapies


The more thorough understanding of signaling pathways implicated in tumor cell proliferation and angiogenesis underlining the pathogenesis of NETs has prompted the application of small molecules targeting these pathways such as everolimus and sunitinib.


Everolimus

Based on preliminary observations, a phase II clinical trial conducted by J. Yao at the MD Anderson Cancer Center studied everolimus, an oral inhibitor of mTOR, and octreotide LAR in advanced low-grade neuroendocrine carcinomas [115]. A total of 60 evaluable patients were treated in two cohorts; patients received either everolimus at 5 or 10 mg/day. Out of the 60 treated patients, 30 had carcinoid and 30 islet cell tumors. Sixty-five percent of the patients were in progression at the time of study entry. Overall, 12 (20 %) patients were reported to have partial response (PR; 4 carcinoids and 8 islet cell tumors, respectively), 43 (72 %) had stable disease (SD; 25 in carcinoids and 18 in islet cell, respectively), and 5 (8 %) had progressive disease (PD; 1 in carcinoids and 4 in islet cell, respectively). Overall median progression-free survival (PFS) was 59 weeks (64 weeks in carcinoids and 50 in islet cell tumors, respectively). Median OS was not reached, with a 2-year survival rate of 78 %. The combination of everolimus and octreotide LAR 30 mg appears to have been well tolerated. The most common toxicity reported was mucositis. Grade 3/4 toxicities observed included anemia, thrombocytopenia, aphthous ulcer, diarrhea, edema, fatigue, hypoglycemia, nausea, pain, and rash.

Another phase II open-label, parallel group study was conducted in patients with advanced PNETs. Patients received either everolimus 10 mg daily as monotherapy (stratum 1; n = 115) or everolimus 10 mg daily in combination with ≤30 mg octreotide LAR q 28 days (stratum 2; n = 45), based on prior octreotide LAR treatment [114]. In stratum 1, there were 11 patients with PRs (9.6 %), 78 patients with SD (67.8 %), and 16 patients with PD (13.9 %); median progression-free survival was 9.7 months. In stratum 2, there were two patients with PRs (4.4 %), 36 patients (80 %) with SD, and no patient with PD; median PFS was 16.7 months.

Following a recently completed phase III trial, the RADIANT-3 [117], that showed activity in a wide range of NETs, everolimus was granted approval by the FDA and EMEA authorities for patients with progressing advanced well-/moderately differentiated PNETs. The RADIANT-3 involved 410 patients with advanced well-/moderately differentiated, radiologically progressing PNETs receiving 10 mg everolimus daily continuously or placebo. The results of this study show prolongation of PFS from 4.6 months in the placebo arm to 11.4 months in the everolimus arm. Grade 1–2 toxicity including stomatitis or aphthous ulceration (64 %), diarrhea (34 %), fatigue (31 %), and infections (23 %) primarily of the upper respiratory tract was mainly observed [117]. Other adverse events include noninfectious pneumonitis and laboratory abnormalities such as anemia, neutropenia, thrombocytopenia, hypercholesterolemia, hypertriglyceridemia, and hyperglycemia [117]. A phase II study investigating the role of another mTOR inhibitor, temsirolimus, was negative for activity in advanced NETs, while the toxic effects were fatigue (78 %), hyperglycemia (69 %), and rash/desquamation (64 %) [24]. A recently published study investigating safety and tolerability of pasireotide LAR in combination with everolimus in patients with advanced NETs showed promising antitumor activity and is further being investigated [14].


Sunitinib

Sunitinib is an oral multi-tyrosine kinases inhibitor that has demonstrated both antiangiogenic and antiproliferative activity by blocking a range of signaling pathways and receptors such as VEGFR-1 to VEGFR-3, stem cell factor receptor, platelet-derived growth factor receptor (PDGFR)-α and PDGFR-β, RET, colony-stimulating factor-1 receptor, and fetal liver tyrosine kinase receptor 3 (FLT-3) [29]. It was originally approved for the treatment of advanced renal cell carcinoma (RCC) and gastrointestinal stromal tumor (GIST) refractory to imatinib treatment. Sunitinib was recently granted authorization for the treatment of advanced well-differentiated progressing PNETs. The exact mechanism underlining the antiproliferative effect of sunitinib is unknown. However, in a NET mouse model, sunitinib has shown an antiangiogenic effect by inhibiting pericytes that support the endothelial cell function [85]. This is most probably achieved by the targeting of PDGFR-β, expressed on pericytes, that drives recruitment and maturation of vessels [6]. The efficacy of sunitinib has been demonstrated in two major clinical studies. The first was a phase II study involving 109 patients with advanced NETs where sunitinib was administered at 50 mg daily for four to six weeks [56]. Of 61 patients with PNETs, 16.7 % had an objective RR, and 68 % a prolonged period of SD. The median time to tumor progression was 7.7 months.

The second was a multinational, randomized, double-blind, placebo-controlled phase III trial comparing sunitinib and best supportive care with placebo and best supportive care. This study recruited patients with advanced well-differentiated PNETs, and sunitinib was delivered at 37.5 mg continuous daily dose [91]. The study was discontinued early as the monitoring committee observed more deaths in the placebo group and a statistically significant difference in progression-free survival (PFS) favoring sunitinib by 6 months [91]. The most frequent adverse events in the sunitinib group were diarrhea (59 %), nausea (44.6 %), vomiting (33.7 %), asthenia (33.7 %), and fatigue (32.5 %). As it results from previous studies, the proactive assessment of other sunitinib-related side effects, such as anorexia, oral changes, hand–foot syndrome and other skin toxicities, thyroid dysfunction, myelotoxicity, hypertension, cardiotoxicity, and thromboembolic events, is critical to ensure optimal treatment benefit by allowing appropriate drug dosing and prolonged treatment periods [51, 107]. Additionally, the objective RR observed in the phase III study was low as it regarded only 9.3 % in the sunitinib group. It is important to identify those patients who may benefit by sunitinib therapy. To this end, a study entitled “Predictive Biomarkers of Response to Sunitinib in the Treatment of Poorly Differentiated NEURO-Endocrine Tumors (NET)” is currently underway in France (clinical trial link: http://​clinicaltrials.​gov/​show/​NCT01215578).


18.5.2.4 Chemotherapy


Over the last three decades, the applied cytotoxic agents for NETs included DNA-damaging agents such as the antimetabolite 5-fluorouracil (5-FU); the alkylating agents streptozotocin (STZ), dacarbazine (DTIC), and temozolomide (TMZ); the topoisomerase inhibitors doxorubicin (Dox) and etoposide; and the DNA crosslinker platinum derivatives, such as cisplatin (CDDP), carboplatin (CBDCA), and oxaliplatin. Single-agent chemotherapy has not shown any real benefit [69], and the newer chemotherapy regimens based on the combination of these active drugs show low RRs, which set the need to improve the results of the medical treatment of these malignancies [108].

Chemotherapy has heterogeneous results in NET patients mainly due to the large variability of biological features among the disease subgroups. For example, chemotherapy in functioning advanced NETs has limited efficacy with a RR of 15 % [102], whereas PNETs have a RR of 30–40 % although with significant toxicity [26, 67]. One of the earliest observations regarding these tumors was that differentiation grade represents one of the most important predictive markers of tumor response to chemotherapy. Patients with metastatic disease of well- and moderately differentiated NF-PNETs are expected to respond better to STZ-based and targeted therapy, whereas those of high-grade NF-PNETs with Ki-67 LI above 20 % are likely to benefit from platinum-based treatment. More specifically, an early report from the Mayo Clinic suggested that anaplastic NETs independently of the primary site location are responsive to combination chemotherapy including etoposide and cisplatin [70]. Conversely, well-differentiated tumors were unresponsive to this regimen [70]. Overall, the population with anaplastic disease had an overall RR of 67 %, a median time to progression (TTP) of 8 months and a median survival of 19 months. Although toxicity of this regimen was severe with vomiting, leukopenia, thrombocytopenia, anemia, alopecia, and neuropathy, the efficacy of this regimen remains unsurpassed by any other treatment until today as there is no unequivocal evidence of survival improvement. Similarly to the grade of differentiation, recent evidence suggests that a careful evaluation of the mitotic index and the Ki-67 assessment can provide further information on the overall prognosis and the degree of response to systemic cytotoxic treatment. For instance, highly proliferative (Ki-67 >30 %) tumors with aggressive behavior such as the PNETs are more sensitive to cytotoxic therapy with cisplatin/etoposide [5]. On the other hand, tumors originating from the small intestine have most commonly a low mitotic index and demonstrate poor response to cytotoxic chemotherapy [5].

One of the first tested combinations was STZ/5-FU in the 1970s and remained a standard treatment until now [68]. There have been very few phase III trials in NETs with conflicting results mainly due to the heterogeneity of the population taken in consideration. The first comparing 5-FU/STZ to Dox showed no statistically significant difference between the two arms [27], and the second comparing 5-FU/Dox to 5-FU/STZ showed the latter being statistically significant better [102]. For PNETs, there have been two phase III trials, one comparing STZ to STZ/5-FU that found no difference between the two arms [69] and a second one comparing chlorozotocin, STZ/5-FU, and STZ/Dox showing a statistically significant benefit in the last arm [71]. During the past decade, there have been few chemotherapy studies in advanced well- and moderately differentiated NETs with encouraging results: (a) with STZ and Dox [21]; (b) with STZ, 5-FU, and Dox [54]; (c) with 5-FU, epirubicin, and dacarbazine [110]; and (d) with STZ, 5-FU, and cisplatin [106]. For the poorly differentiated NETs, the most effective combination chemotherapy remains cisplatin with etoposide [45]. Interestingly, it has been recently reported that gastroenteropancreatic carcinomas with Ki-67 <55 % had a lower RR to platinum-based chemotherapy while having a better prognosis compared with those having Ki-67 >55 % [100]. As a result, it is expected that patients with Ki-67 <55 % may respond to different chemotherapy schedules such as temozolomide, but this needs to be further investigated in the context of a clinical trial [53].

The very high response rates reported in the first studies with STZ-based chemotherapy were criticized since they derived in part from the historical use of nonstandard response criteria. Furthermore, STZ-related toxicity and not manageable schedule limited its use and prompted to newer and better manageable agents like TMZ.

An interesting novel combination chemotherapy is an oral regimen with capecitabine and TMZ that has been shown to exert high activity and low toxicity both in well-differentiated [101] and poorly differentiated neuroendocrine tumors [112]. However, a larger study comparing this regimen with STZ-based therapy or TMZ monotherapy is missing.

Based on the findings of a study assessing retrospectively the triplet capecitabine, STZ, and CDDP, the National Cancer Research Network approved NET01, a randomized phase II study comparing two chemotherapy regimens, capecitabine/STZ and capecitabine/STZ/cisplatin, in unresectable metastatic NETs of the foregut and pancreas and NETs of unknown primary site [106].

Another interesting approach with promising efficacy is the combination of targeted agents with chemotherapy. One such doublet is TMZ and the antiangiogenic agent bevacizumab that has been explored in two different TMZ schedules, a standard [15] and a low continuous dose [52]. A second doublet is TMZ with everolimus that has been tested in 40 patients with PNETs and is shown to achieve a PR in 40 % and a PFS of 15.4 months [13]. However, further studies evaluating the efficacy of these combination therapies compared to TMZ alone are warranted.


18.5.3 Peptide Receptor Radionuclide Therapy


Peptide receptor radionuclide therapy (PRRT) is a targeted radiation therapy involving an intravenously administered radiolabeled somatostatin analog designed to bind on type 2 SSTR overexpressed on PNET cells and deliver a direct and a bystander lethal effect.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Nov 3, 2016 | Posted by in NEUROLOGY | Comments Off on Nonfunctioning Pancreatic Neuroendocrine Tumors

Full access? Get Clinical Tree

Get Clinical Tree app for offline access